Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

Ann. N.Y. Acad. Sci.

ISSN 0077-8923

A N N A L S O F T H E N E W Y O R K A C A D E M Y O F SC I E N C E S
Issue: Annals Meeting Reports

Behavioral epigenetics
Barry M. Lester,1 Edward Tronick,2,3 Eric Nestler,4 Ted Abel,5 Barry Kosofsky,6 Christopher W.
Kuzawa,7 Carmen J. Marsit,8 Ian Maze,9 Michael J. Meaney,10 Lisa M. Monteggia,11 Johannes
M. H. M. Reul,12 David H. Skuse,13 J. David Sweatt,14 and Marcelo A. Wood15
1
Departments of Psychiatry and Human Behavior and Pediatrics, Warren Alpert Medical School, Brown University, Women
and Infants Hospital, Providence, Rhode Island. 2 Department of Psychology, University of Massachusetts, Boston,
Massachusetts. 3 Child Development Unit, Children’s Hospital Boston, Harvard Medical School, Boston, Massachusetts.
4
Mount Sinai Brain Institute, Department of Neuroscience, Mount Sinai School of Medicine, Fishberg Department of
Neuroscience, Mount Sinai School of Medicine, New York, New York. 5 Department of Biology, University of Pennsylvania,
Philadelphia, Pennsylvania. 6 Divsion of Pediatric Neurology, New York-Presbyterian Hospital/Weill Cornell Medical Center,
New York, New York. 7 Institute for Policy Research, Northwestern University, Evanston, Illinois. 8 Department of Pathology and
Laboratory Medicine, Brown University, Providence, Rhode Island. 9 The Rockefeller University, Laboratory of Chromatin
Biology and Epigenetics, New York, New York. 10 Departments of Psychiatry, Neurology, and Neurosurgery, McGill University,
Montreal, Quebec, Canada. 11 Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas.
12
Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, United Kingdom.
13
Behavioural and Brain Sciences Unit, University College of London, Institute of Child Health, London, United Kingdom.
14
Department of Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama. 15 Department of Neurobiology
and Behavior, University of California, Irvine, California

Address for correspondence: Barry Lester, Ph.D., Brown Center for the Study of Children at Risk, Women and Infants Hospital,
101 Dudley St., Providence, RI 02908. Barry_Lester@Brown.edu

Sponsored by the New York Academy of Sciences, the Warren Alpert Medical School of Brown University and the
University of Massachusetts Boston, “Behavioral Epigenetics” was held on October 29–30, 2010 at the University of
Massachusetts Boston Campus Center, Boston, Massachusetts. This meeting featured speakers and panel discussions
exploring the emerging field of behavioral epigenetics, from basic biochemical and cellular mechanisms to the epige-
netic modulation of normative development, developmental disorders, and psychopathology. This report provides
an overview of the research presented by leading scientists and lively discussion about the future of investigation at
the behavioral epigenetic level.

Keywords: behavior; epigenetics; chromosome; gene regulation; transcription; methylation

epigenetics. Behavioral epigenetics was described as


Background and perspectives
the application of the principles of epigenetics to
What is behavioral epigenetics? the study of physiological, genetic, environmental,
Barry M. Lester (Alpert Medical School of Brown and developmental mechanisms of behavior in hu-
University) introduced the topic of the conference, man and nonhuman animals. Investigations typi-
behavioral epigenetics, by describing research on the cally focus at the level of chemical changes, gene
developmental origins of adult diseases, suggesting expression, and biological processes that underlie
that the fetus is actually making adaptations through normal and abnormal behavior. This includes how
programming to “prepare” for the postnatal en- behavior affects and is affected by epigenetic pro-
vironment in response to environmental signals. cesses. Interdisciplinary in its approach, it draws
These effects are due, in part, to epigenetic mech- on sciences, such as neuroscience, psychology and
anisms, raising the fascinating question of whether psychiatry, genetics, biochemistry, and psychophar-
these mechanisms can also explain behavioral out- macology. Whereas there are thousands of studies of
comes, thus providing an example of the kind of epigenetics that have been conducted over the last 40
research that could lead to a new field—behavioral years, the application of epigenetics to the study of

doi: 10.1111/j.1749-6632.2011.06037.x
14 Ann. N.Y. Acad. Sci. 1226 (2011) 14–33 
c 2011 New York Academy of Sciences.
Lester et al. Behavioral epigenetics

Figure 1. The figure shows the 96 articles on behavioral epigenetics grouped by the behavioral construct studied and the genes
that were studied in each of the behavioral construct categories.

behavior is just beginning. A literature search of ci- methylation), ATPase-containing protein com-
tations found only 96 articles to date on behavioral plexes that move histone oligomers along a strand
epigenetics (see Appendix). These articles were an- of DNA, methylation of DNA, and the binding of
alyzed according to the behavioral construct that numerous transcription factors and transcriptional
was studied (e.g., substance use, psychiatric ill- coactivators and corepressors, all of which act in
ness, learning/memory, neurodevelopment, parent- a concerted fashion to determine the activity of a
ing, stress, and neurodegenerative disorders), the given gene. Epigenetic regulation is crucial for ner-
species studied (e.g., human, mouse, rat), the tissue vous system development. Specifically, it can help
that was analyzed (e.g., brain, blood), the epigenetic elucidate how genes are affected by environmen-
mechanisms that were studied (e.g., methylation, tal stimuli, including several common mental retar-
histone modifications), and the particular genes in- dation syndromes and related neurodevelopmen-
vestigated (Fig. 1). For example, in relation to par- tal disorders that are caused by abnormalities in
enting, the most commonly studied genes were the chromatin-remodeling mechanisms.
glucocorticoid receptor and FOS genes. The presen- Epigenetic regulation also occurs in the mature,
tation concluded with cautionary notes about the fully differentiated brain and provides unique mech-
unique issues involved in the study of behavioral anisms that may underlie the stable changes in
epigenetics in humans. gene expression under both normal conditions (e.g.,
learning and memory) and in several pathological
Epigenetics: basic processes and states (e.g., depression, drug addiction, schizophre-
mechanisms nia, and Huntington’s disease, among others). In
Eric Nestler (Mount Sinai School of Medicine) pre- some rare cases (e.g., gene imprinting), epige-
sented an overview of basic epigenetic processes netic modifications can be transmitted to offspring,
and mechanisms.1,2 A broad perspective of epige- which raises the possibility that behavioral experi-
netics includes any structural adaptation in chro- ence in adult life might influence gene expression in
mosomal regions that mediate altered rates of gene subsequent generations. However, there has not yet
transcription. Epigenetic regulation, also known been definitive evidence for epigenetic transmission
as chromatin remodeling, in neurons, describes a of behavioral experience. While work on epigenetic
process where the activity of a particular gene is mechanisms in the brain is still in early stages, it
controlled by the structure of chromatin in that promises to improve our understanding of brain
gene’s proximity (Fig. 2). Chromatin remodeling plasticity, the pathophysiology of neuropsychiatric
is complex, involving multiple covalent modifica- disorders, and may lead to the development of fun-
tions of histones (e.g., acetylation, phosphorylation, damentally new treatments for these conditions.

Ann. N.Y. Acad. Sci. 1226 (2011) 14–33 


c 2011 New York Academy of Sciences. 15
Behavioral epigenetics Lester et al.

Figure 2. General scheme of chromatin remodeling. (A) DNA double helix wrapped around an octomer of histone proteins
forming the unit of chromatin, the nucleosome. (B) Chromatin can be conceptualized as existing in two primary structural states:
as active, or open, euchromatin in which histone acetylation opens up the nucleosome to allow binding of the basal transcriptional
complex and other activators of transcription; or as inactive, or condensed, heterochromatin, where all gene activity is permanently
silenced. In reality, chromatin exists in a continuum of several functional states (active, permissive, repressed, and inactivated).
Enrichment of histone modifications, such as acetylation (A) and methylation (M) at histone N-terminal tails and related binding
of coactivators (Co-Act) or repressors (Rep), to chromatin modulates the transcriptional state of the nucleosome.

Epigenetics, intergenerational inertia, and that organisms must cope with everything from very
human adaptation rapid and acute fluctuations (e.g., overnight fast
Christopher W. Kuzawa (Northwestern University) followed by breakfast) to chronic conditions that
explored the importance of the dynamic nature of change only gradually (e.g., ice ages or migrating to a
epigenetic change as a means by which organisms new environment). A range of adaptive mechanisms
adapt to environmental change.3,4 He emphasized allows human populations to adjust to these various

16 Ann. N.Y. Acad. Sci. 1226 (2011) 14–33 


c 2011 New York Academy of Sciences.
Lester et al. Behavioral epigenetics

timescales of change (Fig. 3). Natural selection sifts ples that suggest an ability of the current generation
through the gene pool to select gene variants well to entrain development to maternal characteristics
suited to the most stable features of local ecologies. that reflect her cumulative experiences include the
Rapid and reversible homeostatic processes lie at setting of infant growth rate to breast milk leptin
the other extreme, maintaining internal constancy as a cue of maternal energetic history, and work
against a backdrop of dynamic environmental con- in the Philippines suggesting that fetal growth may
ditions, such as food intake or psychosocial stress. be calibrated to a woman’s cumulative nutritional
He noted the adaptive importance of developmental experiences across her lifetime. In both examples,
plasticity or the capacity grounded in epigenetic and offspring developmental biology is not sensitive to
other changes that allows a single genome to create potentially transient (and thus unreliable) condi-
a range of possible traits in interaction with the en- tions during the brief period of pregnancy or lacta-
vironment (e.g., growing larger lungs when raised tion. Rather, the maternal resources and signals that
at high altitude). Because organisms only develop are transferred to offspring may be more integrative
once, changing development in response to envi- and cumulative in nature and, thus, potentially pro-
ronmental conditions is generally an irreversible vide a more reliable basis for adaptive adjustment.
process; thus, developmental plasticity is a suitable Kuzawa hypothesized that the timing of early sensi-
mode of adaptation to environmental features that tive periods, during which many epigenetic settings
are too chronic to be buffered by homeostasis, but are established, may be more than accidental, but
that are also too transient for genetic adaptations to reflect the evolution of a conduit of sorts, allowing
consolidate around. nongenomic information to be transferred between
Kuzawa pointed out that many documented ex- generations.
amples of epigenetic sensitivity involve the adoption
Learning and memory
of stable changes in gene regulation in response to
experiences during limited, early stages of devel- The second session, moderated by J. David Sweatt
opment (sensitive periods). Might it make adaptive (University of Alabama at Birmingham), served as
sense for a long-lived species like humans to commit an overview of the roles for epigenetic mechanisms
to a strategy for life so early in the life cycle? Lim- in long-term learning and memory processes and
iting epigenetic sensitivity to early developmental highlights one of the most exciting contemporary
windows may, in fact, create opportunities to ad- areas in the behavioral epigenetics field. The ses-
just biology to more reliable environmental cues in sion was comprehensive in scope, covering cogni-
the form of the mother’s own phenotype. Exam- tion and behavior, synaptic function and cellular

Figure 3. The timescales of human adaptability. Light gray, more rapidly responsive/less durable; black, slowest to respond/most
durable. Epigenetic changes contribute to multiple modes of adaptation, including developmental and intergenerational processes
that allow adjustment to gradual environmental change occurring on a decadal or multigenerational timescale. Modified after
Kuzawa’s work.4

Ann. N.Y. Acad. Sci. 1226 (2011) 14–33 


c 2011 New York Academy of Sciences. 17
Behavioral epigenetics Lester et al.

Figure 4. Schematic representation of epigenetic modifications. (A) In the nucleus, DNA coils and condenses around histones.
Each octameric histone core contains two copies each of histones H2A, H2B, H3, and H4. The DNA–protein complex is referred to
as chromatin. (B) The DNA-histone interaction occurs at the N-terminal tail of a histone, where, for example, on the H3 N-terminal
tail, there are several sites for epigenetic marking via acetylation, methylation, and phosphorylation. (C) In and around gene
promoters that are rich in cytosine-guanine nucleotides (CpG islands), methyl groups are transferred to CpG sites. This process,
called DNA methylation, is catalyzed by a class of enzymes known as DNA methyltransferases.

plasticity, biochemical signaling mechanisms, and hypothesis that DNA methylation marks can be
molecular epigenetic mechanisms. The work de- modified in response to an organism’s experience
scribed largely emphasized the specific epigenetic and that these marks play a role in dynamically reg-
mechanisms of histone posttranslational modifica- ulating the gene transcription supporting synap-
tion and DNA methylation. tic plasticity and long-term memory formation and
maintenance (Fig. 4).
Epigenetic mechanisms in memory formation Sweatt’s presentation also described several pieces
Sweatt addressed the idea that conservation of epi- of evidence supporting the idea that DNA methy-
genetic mechanisms for information storage rep- lation plays a role in memory function in the
resents a unifying model in biology, with epige- adult central nervous system (CNS).6 Thus, he
netic mechanisms being used for cellular memory at described how general inhibitors of DNA methyl-
levels from behavioral memory to development to transferase (DNMT) activity alter DNA methyla-
cellular differentiation.5,6 As background, Sweatt tion in the adult brain and alter the DNA methyla-
discussed how DNA methylation and histone mod- tion status of the plasticity-promoting genes reelin
ifications are the two most extensively investigated and bdnf. Additional studies demonstrated that de
epigenetic mechanisms. As Sweatt described, until novo DNMT expression is upregulated in the adult
recently it was thought that once laid down, these rat hippocampus after contextual fear conditioning
epigenetic marks would remain unchanged for the and that blocking DNMT activity blocks contex-
lifetime of the organism; recent studies, however, in- tual fear conditioning. In addition, results were pre-
cluding those from the Sweatt laboratory, have chal- sented demonstrating that fear conditioning is as-
lenged this view. Nevertheless, it is clear that DNA sociated with rapid methylation and transcriptional
methylation and attendant changes in chromatin silencing of the memory suppressor gene protein
structure are capable of self-regeneration and self- phosphatase 1 (PP1) and demethylation and tran-
perpetuation, necessary characteristics for a stable scriptional activation of the plasticity gene reelin.
molecular mark. Thus, Sweatt discussed the broad These findings have the surprising implication that

18 Ann. N.Y. Acad. Sci. 1226 (2011) 14–33 


c 2011 New York Academy of Sciences.
Lester et al. Behavioral epigenetics

Figure 5. HDAC3 modulates memory formation in a Nr4a2-dependent manner. (A) HDAC3-FLOX mice with dorsal hippocampal
deletion of Hdac3 exhibit significantly enhanced long-term memory as compared to wild-type littermates (both groups received
RISC free control). In contrast, siRNA-Nr4a2–treated HDAC3-FLOX mice exhibit no enhanced memory. (B) qRT-PCR shows that
siRNA against Nr4a2 significantly reduces Nr4a2 expression in both HDAC3-FLOX and wild-type littermates.

both active DNA methylation and active demethy- ing and memory are histone-modifying enzymes,
lation might be involved in long-term memory con- especially histone acetyltransferases (HATs) and his-
solidation in the adult CNS. tone deacetylases (HDACs) (Fig. 5).
Finally, a recent series of studies were described5 In the first part of his talk, Wood presented his
that found that the bdnf gene locus is also sub- lab’s research in examining the role of the CREB-
ject to memory-associated changes in DNA methy- binding protein (CBP), a potent HAT and transcrip-
lation, and, moreover, that this effect is regulated tional coactivator, in long-term memory. One lim-
by the NMDA receptor. Data were also presented itation in studying the role of CBP in learning and
indicating that neuronal DNMT-deficient animals memory has been the lack of genetically modified
have deficits in contextual fear conditioning, the mice with sufficient spatial and temporal regulation.
Morris maze learning task, and hippocampal long- The Wood lab used genetically modified CBP-FLOX
term potentiation (LTP). Overall, Sweatt concluded mice, in combination with adeno-associated virus
that DNA methylation is dynamically regulated in (AAV)–expressing Cre recombinase, to generate ho-
the adult CNS in response to experience and that mozygous focal Cbp deletions in only area CA1 of
this cellular mechanism is a crucial step in memory the dorsal hippocampus. This novel approach re-
formation. sulted in the necessary spatial restriction to study
the role of CBP in one brain region and its effect
Chromatin-modifying enzymes in long-term on long-term memory; additionally, it provided the
memory temporal restriction to study a homozygous deletion
In the second presentation of the session, Marcelo of Cbp in adult mice, which avoids confounds from
A. Wood (University of California, Irvine) dis- developmental or performance issues. The Wood
cussed the role of chromatin-modifying enzymes in lab found that homozygous deletions of Cbp re-
regulating gene expression required for long-term sulted in hippocampus-dependent long-term mem-
memory formation. Why are chromatin-modifying ory impairments associated with decreased levels of
enzymes needed to regulate gene expression? A sim- specific histone modifications and decreased gene
plistic answer comes from the level that compaction expression.7
genomic DNA undergoes when being compressed In the second part of his talk, Wood presented
to fit into a nucleus. Genomic DNA is two me- research examining the role of a specific HDAC in
ters in length, yet has to fit into a six-micron nu- long-term memory formation. To date, the func-
cleus, and, thus, must undergo an approximately tion of HDAC3, one of the most highly expressed
10,000-fold compaction. This generates an access class I HDACs in the brain, has never been ex-
and indexing problem, which is solved in part by amined. Again, using AAV-expressing Cre recombi-
chromatin-modifying enzymes. The best-studied nase and HDAC3-FLOX genetically modified mice,
chromatin-modifying enzymes in the field of learn- the Wood lab demonstrated that HDAC3 is a key

Ann. N.Y. Acad. Sci. 1226 (2011) 14–33 


c 2011 New York Academy of Sciences. 19
Behavioral epigenetics Lester et al.

negative regulator of long-term memory forma- as a site of action for glucocorticoids in stress-related
tion in the hippocampus. Homozygous deletion memory formation.
of Hdac3 in area CA1 of the hippocampus led to In his presentation, Reul showed that the hor-
enhanced long-term memory associated with in- mone’s action required distinct epigenetic modifi-
creased levels of specific histone modifications and cations of the chromatin: the phosphorylation of
increased gene expression. Similar results were ob- serine10 (S10), in combination with the acetylation
served when an HDAC3 selective inhibitor, called of lysine14 (K14) of histone H3 (H3S10p-K14ac),
RGFP136, was site specifically delivered to the dorsal leading to the induction of the immediate-early
hippocampus. Together, the genetic and pharmaco- genes c-Fos and Egr-1 in dentate gyrus granule neu-
logic data demonstrated that HDAC3 is a negative rons in rats and mice in vivo.11,12 As the gluco-
regulator of long-term memory formation.8 corticoid receptor (GR) cannot bring about these
In summary, Wood posited that HDACs represent histone modifications directly, Reul suggested that
a type of molecular brake pad that is normally en- the GR acts indirectly via interaction with other sig-
gaged but transiently removed by sufficient activity- naling molecules. More specifically, he postulated
dependent signaling to regulate the transcription that GR interacts with the NMDA receptor-activated
required for long-term memory formation.9 Wood ERK (extracellular signal-regulated kinase) MAPK
concluded by suggesting that this process may rep- (mitogen-activated protein kinase) signal pathway,
resent a molecular mechanism to explicate why we which has a marked role in learning and memory
do not encode everything we experience into long- processes (Fig. 6; Refs. 12 and 13). Supporting this
term memory. Moreover, impaired function of these notion, Reul presented unpublished in vivo data
molecular brake pads may be associated with disor- clearly showing that in activated dentate granule
ders including drug addiction and posttraumatic neurons, that is, those exhibiting phosphorylated
stress disorder. ERK1/2 (pERK1/2), GRs are required to activate
the downstream histone-modifying enzymes MSK1
Signaling and epigenetic mechanisms (mitogen and stress-activated kinase 1), and Elk-1
in stress-related memory formation (Ets-like protein-1) (Fig. 6; Refs. 12–14).
In the final presentation, Johannes (Hans) Reul Reul went on to describe pMSK1, a kinase that
(University of Bristol, UK) presented a novel mech- can phosphorylate histone H3 at serine10, whereas
anism that may explain why we make such strong pElk-1 binds the HAT p300, which can acetylate his-
memories of psychologically stressful and emotional tone tails. Further, showing a series of immunoflu-
events in our lives. The mechanism he proposed orescence images, he demonstrated that, during the
involves crosstalk between different signaling path- consolidation phase of memory formation, all par-
ways influencing epigenetic processes in neurons of ticipating signaling molecules (pERK1/2, pMSK1,
the hippocampus, a limbic brain region involved in pElk-1), modified histone molecules (H3S10p-
learning and memory. Stressful events, for example, K14ac), and induced intermediate-early gene
a domestic dispute or a job interview, or in ani- products (c-Fos, Egr-1) can be found in the same
mals, an attack by a predator, evoke the secretion of dentate gyrus granule neurons.12,13 Furthermore,
glucocorticoid hormones from the adrenal gland. he showed that blocking GRs led to a substan-
Classically, these hormones regulate metabolic and tially decreased formation of pMSK1 and pElk-1,
other physiological processes that enable an individ- but not pERK1/2, in dentate granule neurons af-
ual to cope with the challenge in the best possible ter forced swim stress.12 He concluded that stress-
way. ful events are strongly encoded into memory be-
Reul reported, however, that research spanning cause of the marked activating role of GRs on ERK
the last 25 years has provided evidence that gluco- MAPK, signaling to the chromatin in dentate gyrus
corticoids secreted during a psychologically stressful neurons (Fig. 6; Ref. 12). These findings may be of
challenge enhance the consolidation of memories significance for stress-related psychiatric disorders,
related to the event—a long-standing observation such as major depression and anxiety, including
that has remained unexplained until now. A finding PTSD.
made in the 1980s by de Kloet’s group10 pointed to The formal presentations were followed by a
the dentate gyrus, the gateway of the hippocampus, wide-ranging and lively discussion of the roles

20 Ann. N.Y. Acad. Sci. 1226 (2011) 14–33 


c 2011 New York Academy of Sciences.
Lester et al. Behavioral epigenetics

Figure 6. Glucocorticoid hormones, secreted as a result of a stressful event, enhance the consolidation of behavioral responses,
including memories related to the event. Until recently, the underlying mechanisms of these effects were unknown. Recent
work of the Reul group at the University of Bristol shows that glucocorticoids act by binding to glucocorticoid receptors (GRs)
that interact with the NMDA receptor-activated ERK1/2/MSK1-Elk-1 signaling pathway enhancing the formation of epigenetic
modifications (i.e., the serine10 phosphorylation and lysine14 acetylation in histone H3 (H3S10p-K14ac)) and the induction of the
neuroplasticity-associated immediate-early genes c-Fos and Egr-1 in sparsely distributed mature dentate gyrus neurons. Evidence
has been accumulating that these signaling, epigenetic, and genomic phenomena are of critical importance for the consolidation of
memories related to the endured stressful event.

and regulation of epigenetic mechanisms in long- Alterations of DNA methylation, growth


term synaptic plasticity and behavioral memory restriction, and infant neurobehavior
in vivo. The first talk was by Carmen J. Marsit (Brown Uni-
versity) and focused on altered epigenetic marks
within the placenta. Epidemiological studies iden-
Development
tify variations in birth weight as a predictor of health
This section, chaired by Edward Tronick (Univer- over the lifespan, including the risk for neuropsy-
sity of Massachusetts Boston and Children’s Hospi- chiatric disorders.15 Marsit discussed a novel way
tal Boston), focused on the emerging environmental in which to consider the effects of the intrauter-
epigenetics hypothesis, which suggests that environ- ine environment on infant neurodevelopment in
mental signals operate during early development to human populations, focusing on how differences
alter epigenetic marks across the genome, thus in- in DNA methylation at specific genomic regions
fluencing neural development and function. in the human placenta are associated with infant

Ann. N.Y. Acad. Sci. 1226 (2011) 14–33 


c 2011 New York Academy of Sciences. 21
Behavioral epigenetics Lester et al.

neurobehavior. The placenta acts as the master reg- to maternal behaviors (see Meaney presentation
ulator of the intrauterine environment, not only below).
through nutrients, gas, water, and waste exchange, Marsit also showed that these effects were most
but also through the production of pregnancy- pronounced among infants of normal weight for
related hormones, proteins, and growth factors, in- gestational age, suggesting that there may be normal
cluding neuropeptide hormones analogous to those variability in placental methylation that accounts
produced by the hypothalamus and pituitary. Fi- for variation in infant neurobehavior. As Marsit
nally, the placenta also acts as a barrier commonly expands his studies of the role of the intrauterine
metabolizing maternal hormones to inactive forms environment captured in the placenta epigenome,
and, thus, stabilizing the intrauterine endocrine en- links between the methylation of key genes involved
vironment. Such considerations have led to the con- in HPA axis control and infant neurobehavior, and
cept that the placenta acts as the “third brain” by associations between genome-wide profiles of DNA
linking the developed maternal physiological state methylation and infant neurodevelopment are being
with the developing fetus. pursued. These studies are of particular importance
Importantly, placental gene expression is subject as multiple environmental exposures, such as nutri-
to environmental regulation. Marsit’s group con- ent deprivation, are known to affect infant growth
sidered how changes to the patterns of DNA methy- and are associated with an increased risk for neu-
lation in the placenta may alter the function of the rocognitive conditions, including attention deficit
placenta in these critical roles and, in turn, how these hyperactivity disorder (ADHD).
alterations manifest in neurobehavioral phenotypes
in infants, characterized using the well-established Epigenetic alterations and exposure
Neonatal Intensive Care Unit Network Neurobehav- to cocaine in utero
ioral Scales (NNNS). Barry Kosofsky (Weill Cornell Medical College) dis-
Marsit highlighted work linking patterns of DNA cussed how developmental brain disorders and the
methylation in the placenta to the intrauterine en- consequences of prenatal exposure to drugs of abuse
vironment represented by infant growth, showing (cocaine, in particular) are associated with sustained
strong and significant associations between profiles changes in CNS gene expression and have last-
of DNA methylation, identified using genome-wide ing consequences for brain structure and function
array-based approaches, and infant birth weight.16 (Fig. 8).17 Prenatal exposure to toxins, including
He went on to demonstrate that increasing methy- substances of abuse, is associated with developmen-
lation of the human GR 1F was strongly and sig- tal effects in children. Kosofsky suggested that such
nificantly associated with decreased measures of aberrant effects might be considered “molecular
infant attention on the NNNS (Fig. 7). Impor- malformations,” leading to conditions where neural
tantly, the methylation of an analogous receptor signaling pathways are rendered dysfunctional. Such
(rat exon 17 ) in rat pup hippocampus has linked molecular changes may “feed forward” to produce

Figure 7. Association between greater than median glucocorticoid receptor exon 1F methylation and infant attention score is
specific to nongrowth restricted infants.

22 Ann. N.Y. Acad. Sci. 1226 (2011) 14–33 


c 2011 New York Academy of Sciences.
Lester et al. Behavioral epigenetics

Figure 8. Epigenetic mechanisms underlying persistent alterations on promoters of genes involved in neuronal plasticity.

alterations in the behavioral repertoire of affected demonstrating that mice exposed to cocaine pre-
infants, children, and young adults—changes that natally demonstrated impaired spontaneous recov-
are sculpted by environmental interactions. Kosof- ery of extinction, a form of learning that relies on
sky’s research explores the hypothesis that the result- the mPFC. The prenatally cocaine-exposed animals
ing molecular maladaptations are, in part, mediated showed a decrease in the binding of MeCP2 to the
by epigenetic mechanisms. promoter of exons I and IV of the bdnf gene, which
Kosofsky presented data from a mouse model was associated with decreased mRNA expression
of transplacental cocaine exposure. These findings of those transcripts in mPFC, likewise suggesting
suggest that changes are expressed in a gene-specific, an epigenetic mechanism underlying the behavioral
region-specific, and time-specific fashion; when ap- alterations. These findings are consistent with the
parent in the medial prefrontal cortex (mPFC), these presentation of David Sweatt in an earlier session
changes appear to result in altered structural and on epigenetic mechanisms for learning and mem-
functional maturation of that brain region. When ory that highlighted the importance of epigenetic
compared with control animals (i.e., mice with no regulation of the bdnf gene for fear conditioning.
prenatal drug exposure), the cocaine-exposed mice The implication from these findings is that perina-
showed a differential pattern of performance on a tal environmental conditions might determine the
social interaction (SI) task: increased SI relative to capacity for neural plasticity in later life through
controls at P28 (juvenile) and decreased SI relative epigenetic regulation of genes critical for synaptic
to controls as adults. A parallel pattern of expres- remodeling. Kosofsky’s group is currently pursuing
sion of the mRNA for the transcription factor EGR1 “rescue experiments” to further explore the link be-
(also known as NGF-1a and zif 268) was observed in tween the proposed molecular mechanisms in the
mPFC corresponding with these ages. In adult an- animals prenatally treated with cocaine. The find-
imals, changes in EGR1 expression correlated with ings may provide an opportunity for translational
decreased binding of MeCP2 to the EGR1 promoter; benefit regarding the diagnosis and treatment for
the same pattern was not observed at P28. Vari- the offspring of woman who abuse drugs during
ations in MeCP2 occupancy suggest that an epi- pregnancy.
genetic mechanism may underlie changes in gene
expression and behavior. Epigenetic programming by maternal care
Kosofsky presented additional behavioral studies Michael J. Meaney (McGill University) summarized
using an “extinction of conditioned fear” model, previous studies showing that variations in maternal

Ann. N.Y. Acad. Sci. 1226 (2011) 14–33 


c 2011 New York Academy of Sciences. 23
Behavioral epigenetics Lester et al.

Figure 9. Tactile stimulation derived from pup LG increases 5-HT activity at the level of the hippocampus, thus increasing NGFI-A
expression and its association with the exon 17 promoter, which then initiates an alteration of the methylation state of the exon 17
glucocorticoid receptor promoter.

care in the rat, specifically in the frequency of pup mone. T3 increases central 5-HT activity in the rat
licking/grooming (LG), is associated with increased pup, and its administration is sufficient to increase
methylation of the exon 17 GR promoter in the the association of NGFI-A with the exon 17 pro-
hippocampus, decreased hippocampal FR expres- moter. Pup LG from the mother directly increases
sion, and increased hypothalamic-pituitary-adrenal NGFI-A association with the exon 17 promoter, and
(HPA) responses to stress.18,19 Previous work sug- artificial tactile stimulation mimics this effect. These
gests that reversing the effects of differential DNA findings suggest that the tactile stimulation derived
methylation of the exon 17 promoter, in turn, can from pup LG increases 5-HT activity at the level of
reverse the effects of maternal care on hippocampal the hippocampus, thus increasing NGFI-A expres-
FR expression and HPA responses to stress. Meaney sion and its association with the exon 17 promoter,
also presented findings from studies in the post- which then initiates an alteration of the methyla-
mortem human hippocampus showing that a de- tion state of the exon 17 GR promoter (Fig. 9).
velopmental history of child abuse was associated The results are consistent with previous studies in
with an increase in the methylation of the exon 1F vitro showing that overexpression of NGFI-A alters
GR promoter (also see Marsit summary) and de- the methylation of the exon 17 promoter. Interest-
creased GR expression. The focus of the talk con- ingly, NGFI-A also regulates the expression of the
cerned the mechanisms by which the environmen- GAD1 gene that encodes glutamic acid decaroxy-
tal signal, pup LG, might generate the difference lase 1, and maternal care regulates the methylation
in DNA methylation, and gene expression. Meaney of the GAD1 and GAD1 expression in a manner
summarized in vitro and in vivo evidence for the im- comparable to that of the GR. These studies are
portance of serotonin (5-HT)- and 5-HT–induced consistent with earlier reports of alterations in DNA
increases in hippocampal NGFI-A expression for the methylation associated with increased transcription
alterations in the methylation state of the exon 17 factor binding, and suggest that environmental con-
promoter. A shRNA targeting NGFI-A blocks both ditions can directly alter epigenetic states through
the effects of 5-HT on the methylation state of the the activation of intracellular signaling pathways.
exon 17 promoter and effects on GR expression. Pup Meaney also noted important caveats, most notably,
LG provides tactile stimulation of the pup, resulting the importance of identifying the enzyme directly
in an increase in circulating levels of triiodoithyro- responsible for the alteration in the methylation
nine (T3), the most biologically active thyroid hor- state.

24 Ann. N.Y. Acad. Sci. 1226 (2011) 14–33 


c 2011 New York Academy of Sciences.
Lester et al. Behavioral epigenetics

Summary of symposium on development nisms by which drugs of abuse and stress produce
Each of these presentations focused on well- long-lasting alterations in gene expression and be-
established environmental influences, including havior.20 Moreover, identifying common regulatory
pre- and postnatal maternal effects and drugs of mechanisms in cocaine and stress models may aid in
exposure. This research reflects an emerging body the development of therapeutics aimed at alleviating
of science examining epigenetic states as candidate addiction and depression syndromes.
mechanisms that link environmental conditions in
Epigenetic targets in neurodegenerative and
early development with sustained changes in gene
psychiatric disorders
expression and neural development. Predictably, the
Ted Abel (University of Pennsylvania) focused
discussion focused on the enthusiasm for the po-
on another type of crucial histone modification,
tential benefits of interventions targeting epigenetic
namely, histone acetylation, which is generally as-
mechanisms. The speakers noted that the current
sociated with transcriptional activation. Histone
period marks a very early stage for research linking
acetylation is catalyzed by HATs and reversed by
environmental conditions to alterations in gene ex-
HDACs. One of the major HATs present in the brain
pression and brain function. Nevertheless, the trans-
is the transcriptional coactivator, termed CBP. Abel
lational science presented within this symposium
et al. have shown that CBP is involved in synaptic
suggests that epigenetics represents a fruitful area
plasticity in the hippocampus and in specific forms
of research bridging epidemiological findings with
of long-term memory mediated via hippocampal
studies of biological mechanism.
circuits. Thus, mutant mice, in which CBP activ-
Neuropsychiatry ity in neurons is reduced, exhibit deficits in spatial
and contextual memory and in long-lasting forms
Histone methylation in cocaine-induced
of hippocampal synaptic plasticity. A complemen-
behavioral and structural plasticity
tary method to study the role of histone acetylation
Ian Maze presented research from Eric Nestler’s lab-
in synaptic plasticity and memory is to examine the
oratory (Mount Sinai School of Medicine) that di-
effects of HDAC inhibitors, which increase histone
rectly implicates a role for repressive histone methy-
acetylation and transcriptional activation. The Abel
lation, specifically dimethylation of Lys9 on histone
laboratory and other groups have found that ad-
H3 (H3K9me2), in cocaine addiction (Fig. 10).
ministration of an HDAC inhibitor, such as tricho-
Maze et al. have shown that chronic cocaine
statin A, enhances long-term contextual memory
administration to mice reduces global levels of
and facilitates synaptic plasticity via the transcrip-
H3K9me2 in nucleus accumbens (NAc), a key brain
tion factor CREB. Among important target genes
region involved in processing reward and implicated
induced by HDAC inhibition and CREB in the hip-
in addiction.20 In NAc, the reduction in H3K9me2
pocampus are certain nuclear receptor transcription
is mediated through decreased expression of G9a, a
factors that are critical for the enhanced cognitive
histone methyltransferase that specifically catalyzes
ability observed. Histone acetylation may, therefore,
H3K9me2. The repression of G9a, in turn, is me-
provide an epigenetic mechanism for establishing
diated by the cocaine-induced transcription factor,
gene-specific modifications that result in the co-
FosB. Using conditional mutagenesis and viral-
ordinate expression of genes required for long-term
mediated gene transfer, the group found that G9a
memory storage. As well, HDAC inhibitors may pro-
downregulation increases dendritic spine plasticity
vide a novel therapeutic approach to treat the cogni-
of NAc neurons and enhances rewarding responses
tive deficits that accompany many neuropsychiatric
to cocaine by decreasing repressive H3K9me2 at spe-
disorders.
cific target genes and, therefore, increasing those
genes’ expression. Cocaine-induced downregula- Epigenetic mechanisms regulating synapse
tion of G9a and H3K9me2 also promotes an in- function and behavior
dividual’s vulnerability to stressful experiences and Lisa M. Monteggia (The University of Texas South-
the development of depression-like behavioral ab- western Medical Center at Dallas) discussed her lab-
normalities. These findings are consistent with clin- oratory’s studies of loss-of-function mutations in
ical data that drug addiction and depression often the gene methyl-CpG-binding protein-2 (MeCP2)
occur together. This work has defined new mecha- that cause Rett syndrome, a neurodevelopmental

Ann. N.Y. Acad. Sci. 1226 (2011) 14–33 


c 2011 New York Academy of Sciences. 25
Behavioral epigenetics Lester et al.

Figure 10. Mechanism of increased cocaine sensitivity.

disorder characterized by reduced cognitive func- tion of synaptic function. The regulation of these
tion and autism spectrum-like behavioral abnor- two key epigenetic mechanisms by synaptic activity,
malities, among other deficits. and how such alterations affect neurotransmission,
Monteggia et al., along with other groups, have will be critical to elucidate the mechanisms under-
demonstrated that mice lacking MeCP2 exhibit ab- lying Rett syndrome as well as the roles these factors
normal cognitive and social behavior. The group have in basic cellular processes. This work is also
has also demonstrated that loss of MeCP2 de- essential in understanding abnormalities in neuro-
creases excitatory glutamatergic transmission in transmission that underlie Rett syndrome and other
the hippocampus, primarily by reducing glutamate neuropsychiatric disorders.
release. By contrast, no deficit in inhibitory
GABAergic function is seen. The results suggest Epigenetic risk factors in
that some Rett abnormalities are caused by an social-communication disorders
imbalance between excitatory and inhibitory neu- David H. Skuse (University College London in the
rotransmission in particular brain circuits, a possi- UK) discussed genomic imprinting, which involves
bility supported by work from the Monteggia lab- epigenetic modifications that result in differential
oratory. MeCP2, encoded by the X chromosome, gene expression from certain genes (or even entire
functions predominantly by binding to methylated chromosomes) that are of paternal versus maternal
CpG islands in the promoter region of certain genes origin.
and thereby silencing those genes’ expression. This Importantly, Skuse et al. have proposed that
occurs, in part, by forming a protein complex with imprinting of the X-chromosome could result in
HDACs, which also repress gene activation, as noted sexually dimorphic characteristics. This hypothesis
above. The Monteggia laboratory has, therefore, predicts that sexually dimorphic (male) vulnerabil-
started to investigate the coordinated role of histone ity to some neurodevelopmental disorders, such as
deacetylation and DNA methylation in the regula- autism, could occur on the basis of whether the

26 Ann. N.Y. Acad. Sci. 1226 (2011) 14–33 


c 2011 New York Academy of Sciences.
Lester et al. Behavioral epigenetics

silencing of alleles is confined to chromosomes that ified. Yet any or all of these environmental factors,
were either paternally or maternally derived.21 and the animals’ engagement with them, may lead
The X-chromosome is enriched for genes that to epigenetic effects. Indeed the presumption of epi-
are involved in brain function. X-monosomy in hu- genetics is that such factors do and will have effects.
mans results in Turner syndrome, which provides a For example, the stability of epigenetic changes may
model by which these putative mechanisms can be be the result of changes in environmental factors
studied. In general, studies have shown that females that function to stabilize, destabilize, or even reit-
with a single X-chromosome of paternal origin have eratively induce epigenetic changes rather than the
better social communication skills, and are more stability being “inherent” to the epigenetic change
empathic, than those whose single X-chromosome in and of itself. Moreover, the dynamics of change
is maternal in origin.22 Autosomal gene expression need to be better understood; that is, epigenetic
may be regulated by X-linked genes.23 changes related specifically to external environmen-
Skuse’s laboratory has shown in longitudinal tal events in turn can become causal elements that go
studies that these differences persist from childhood on to amplify, stabilize, or inhibit other epigenetic
into adulthood. These human observations have changes. These dynamics have come to be appreci-
more recently been followed-up by studies of X- ated in studies of physiologic systems such as the
monosomic mice. Replicated findings include pref- HPA axis, where physiologic and behavioral feed-
erential expression of alleles from the maternally back and feed-forward loops operating over time are
derived X-chromosome. One particular allele, in- critical to understanding how the organism func-
variably expressed in males, is associated with per- tions. Similar dynamic thinking needs to be intro-
severative behavior.24 No evidence for preferential duced into our models of epigenetic changes.
expression of the paternally derived X-chromosome Tronick noted that most of the models we have
has yet been observed, although recent work from for epigenetic mechanisms related to behavior are
Catherine Dulac’s laboratory at Harvard has pro- models of abnormal processes, such as toxic expo-
vided support for the role of X-linked imprinting in sures or deprivation. We know less about epigenetic
brain development.25 processes that affect normal behavior as was shown
Better understanding the mechanisms of genomic in work of Marsit. Thus much of what we know
imprinting has the potential of providing new in- may be related to aberrant processes that fall out-
sight into the molecular basis of individual variabil- side the range of normal epigenetic processes. For
ity in personality traits as well as features of neu- example, the finding of high levels of methylation
ropsychiatric disorders. early in development could suggest that the timing
of the effects of experience may be critical to under-
Concluding Remarks
standing epigenetic effects on behavior. Moreover,
This conference brought together cutting-edge an- the idea of developmental change as being, in part, a
imal and human research in the emerging field of process related to a release from methylation would
behavioral epigenetics. As exciting as these findings have profound effects for our understanding of de-
are, there are a number of challenges that need to velopment, such as identifying the events and their
be addressed as the field moves forward. Ed Tronick timing that trigger the release from methylation, as
(University of Massachusetts and Harvard Medical well as for therapeutic interventions.
School) pointed out that given what is now known Tronick recognized that at the present time our
about environmental effects that there needs to be ability to specify the chain of causality of epigenetic
as much effort put into characterizing the details of changes in human behavior is limited because of our
the experience of the animal or human and its envi- inability to access brain tissue. One can only hope
ronment as has gone into characterizing molecular that new techniques will be developed that overcome
mechanisms. At the moment experiential and envi- this limitation and that with a better understand-
ronmental “phenotyping” is crude. Even in the best ing of tissue may lead to the finding of “substitutes
animal experimental studies, factors in addition to tissue” and correlated changes in other physiologi-
the study proper, such as housing conditions, events cal systems. For example, epigenetic changes in the
during animal housing, handling regimes, light cy- glucocorticoid receptor detected in plasma or buc-
cles, social contacts, to name a few are not well spec- cal cells accompanied by parallel changes in ACTH,

Ann. N.Y. Acad. Sci. 1226 (2011) 14–33 


c 2011 New York Academy of Sciences. 27
Behavioral epigenetics Lester et al.

CRF or cortisol would strengthen the role of the HPA References


axis. But while we wait for these new innovations
1. Jenuwein, T. & C.D. Allis. 2001. Translating the histone code.
there needs to be an appreciation of the limitations Science 293: 1074–1080.
of human epigenetic work compared to the research 2. Tsankova, N., W. Renthal, A. Kumar & E.J. Nestler. 2007.
done on animals. The state of the art of the two areas Epigenetic regulation in psychiatric disorders. Nature Rev.
is not the same, and applying state-of-the-art animal Neurosci. 8: 355–367.
standards to human work will only limit progress. 3. Kuzawa, C.W. & E.A. Quinn. 2009. Developmental origins of
adult function and health: evolutionary hypotheses. Annu.
Tracing human behavior to epigenetic changes will Rev. Anthropol. 38: 131–147.
be difficult, but we can make every effort to have 4. Kuzawa, C.W. 2005. The fetal origins of developmental plas-
as much detailed characterization of the epigenetic, ticity: are fetal cues reliable predictors of future nutritional
physiologic, behavioral, and environmental links as environments? Am. J. Hum. Biol. 17: 5–21.
5. Miller, C.A. et al. 2010. Cortical DNA methylation maintains
possible and then to fill in the hidden links with
remote memory. Nat. Neurosci. 13: 664–666.
work using animal models. That said, whatever the 6. Miller, C.A. & J.D. Sweatt. 2007. Covalent modification
organism, we need to appreciate the full complexity of DNA regulates memory formation. Neuron 53: 857–
of epigenetic changes. 869.
Certainly, part of the excitement of this confer- 7. Barrett, R.M., M. Malvaez, E. Kramar, et al. 2011. Hippocam-
pal focal knockout of CBP affects specific histone modi-
ence was the coming together of scientists from dif-
fications, long-term potentiation, and long-term memory.
ferent disciplines, such as molecular biologists and Neuropsychopharm. Apr. 20 [Epub ahead of print].
behavioral scientists, who are capable of developing 8. McQuown, S.C., R.M. Barrett, D.P. Matheos, et al. 2011.
models that will elucidate both the hidden links and HDAC3 is a critical negative regulator of long-term memory
the complexity to further advance this relatively new formation. J. Neuorsci. 31: 764–774.
9. McQuown, S.C. & M.A. Wood. 2011. HDAC3 and the molec-
field of behavioral epigenetics.
ular brake pad hypothesis. Learn. Mem. Apr. 16 [Epub ahead
of print].
Acknowledgments 10. De Kloet, E.R., S. De Kock, V. Schild & H.D. Veldhuis. 1988.
Antiglucocorticoid RU 38486 attenuates retention of a be-
The Behavioral Epigenetics conference was pre- haviour and disinhibits the hypothalamic-pituitary adrenal
sented by the New York Academy of Sciences, the axis at different brain sites. Neuroendocrinol. 47: 109–115.
Warren Alpert Medical School of Brown Univer- 11. Bilang-Bleuel, A. et al. 2005. Psychological stress increases
sity, and the University of Massachusetts Boston, histone H3 phosphorylation in adult dentate gyrus gran-
ule neurons: involvement in a glucocorticoid receptor-
and supported in part by the University of Mas-
dependent behavioural response. Eur. J. Neurosci. 22: 1691–
sachusetts Boston and the Life TechnologiesTM 1700.
Foundation (Silver), the Massachusetts Life Sciences 12. Gutierrez-Mecinas, M., A. Collins, X. Qian, et al. 2009.
Center (Bronze), and Bristol-Myers Squibb R&D Forced swimming-evoked histone H3 phospho-acetylation
and Genomatix Software, Inc (Academy Friends). and c-Fos induction in dentate gyrus granule neurons in-
volves ERK1/2-mediated MSK1 and Elk-1 phosphorylation.
Funding for this conference was also made possi-
Soc. Neurosci. Abst. 777: 17.
ble by (i) Grant 1 R13 DA029985-01 from the Na- 13. Reul, J.M.H.M., S.A. Hesketh, A. Collins & M. Gutierrez-
tional Institute on Drug Abuse, the Eunice Kennedy Mecinas. 2009. Epigenetic mechanisms in the dentate gyrus
Shriver National Institute of Child Health and Hu- act as a molecular switch in hippocampus-associated mem-
man Development, the National Institute of Mental ory function. Epigenetics 4: 434–439.
14. Chandramohan,Y., S.K. Droste, J.S. Arthur & J.M.H.M. Reul.
Health, and the National Institutes of Health, Office
2008. The forced swimming-induced behavioural immobil-
of the Director (Barry M. Lester, Principal Investiga- ity response involves histone H3 phospho-acetylation and
tor); (ii) an Independent Medical Education Grant c-Fos induction in dentate gyrus granule neurons via ac-
from AstraZeneca; (iii) March of Dimes Founda- tivation of the N -methyl-D-aspartate/extracellular signal-
tion Grant No. 4-FY10-458; and (iv) an educational regulated kinase/mitogen- and stress-activated kinase sig-
nalling pathway. Eur. J. Neurosci. 27: 2701–2713.
grant from Janssen, a division of Ortho-McNeil-
15. Schlotz, W. & D.I. Phillips. 2009. Fetal origins of mental
Janssen Pharmaceuticals, Inc., administered by health: evidence and mechanisms. Brain Behav. Immun. 23:
Ortho-McNeil-Janssen Scientific Affairs, LLC. 905–916.
16. Filiberto, A.C. et al. 2011. Birthweight is associated with
Conflicts of interest DNA promoter methylation of the glucocorticoid receptor
in human placenta. Epigenetics May 1; 6(5). [Epub ahead of
The author declares no conflicts of interest. print].

28 Ann. N.Y. Acad. Sci. 1226 (2011) 14–33 


c 2011 New York Academy of Sciences.
Lester et al. Behavioral epigenetics

17. Crandall, J.E., H.E. Hackett, S.A. Tobet, et al. 2004. Cocaine man brain associates with childhood abuse.
exposure decreases GABA neuron migration from the gan- Nat. Neurosci. 12: 342–348.
glionic eminence of the cerebral cortex in mice. Cereb. Cort.
7. Oberlander, T.F. 2008. Prenatal exposure to
14: 665–675.
18. Zhang, T.Y., I.C. Hellstrom, R.C. Bagot, et al. 2010. Maternal maternal depression, neonatal methylation of
care and DNA methylation of a glutamic acid decarboxylase human glucocorticoid receptor gene (NR3C1)
1 promoter in rat hippocampus. J. Neurosci. 39: 13130– and infant cortisol stress responses. Epigenetics
13137. 3: 97–106.
19. Meaney, M.J. & A.C Ferguson-Smith. 2010. Epigenetic regu-
8. Vecsey, C.G. 2007. Histone deacetylase in-
lation of the neural transcriptome: the meaning of the marks.
Nat. Neurosci. 11: 1313–1318. hibitors enhance memory and synaptic plastic-
20. Maze, I. et al. 2010. Essential role of the histone methyl- ity via CREB: CBP-dependent transcriptional
transferase G9a in cocaine-induced plasticity. Science 327: activation. J. Neurosci. 27: 6128–6140.
213–216. 9. Pandey, S.C., R. Ugale, H. Zhang, et al. 2008.
21. Skuse, D. 2006. Genetic influences on the neural basis of
Brain chromatin remodeling: a novel mecha-
social cognition. Philos. Trans. R. Soc. Lond. B Biol. Sci. 361:
2129–2141. nism of alcoholism. J. Neurosci. 28: 3729–3737.
22. Skuse, D.H. et al. 1997. Evidence from Turner’s syndrome 10. Kumar, A. et al. 2005. Chromatin remodeling is
of an imprinted X-linked locus affecting cognitive function. a key mechanism underlying cocaine induced
Nature 387: 705–708. plasticity in striatum. Neuron 48: 303–314.
23. Wijchers, P.J. & R. J. Festenstein. 2011. Epigenetic regulation
11. Alarcon, J.M. et al. 2004. Chromatin acety-
of autosomal gene expression by sex chromosomes. Trends
Genet. 4: 132–140. lation, memory, and LTP are impaired in
24. Davies, W. 2010. Genomic imprinting on the X chromo- Cbp/ mice: a model for the cognitive deficit
some: implications for brain and behavioral phenotypes. in Rubinstein-Taybi syndrome and its amelio-
Ann. N.Y. Acad. Sci. 1204(Suppl.): E14–E19. ration. Neuron 42: 947–959.
25. Gregg, C. et al. 2010. Sex-specific parent-of-origin allelic
12. Weaver, I.C.G. et al. 2004. Epigenetic program-
expression in the mouse brain. Science 329: 682–685.
ming by maternal behavior. Nat. Neurosci. 7:
847–854.
13. Weaver, M.J., M. Meaney & M. Szyf. 2006. Ma-
Appendix: ninety-six articles published to
ternal care effects on the hippocampal tran-
date on behavioral epigenetics.
scriptome and anxiety-mediated behaviors in
1. Fischer, A., F. Sananbenesi, X. Wang, et al. the offspring that are reversible in adult-
2007. Recovery of learning and memory is as- hood. Proc. Natl. Acad. Sci. USA 103: 3480–
sociated with chromatin remodeling. Nature 3485.
447: 178–182. 14. Weaver, I.C.G. et al. 2005. Reversal of maternal
2. Mueller, B.R. & T.L. Bale. 2008. Sex-specific programming of stress responses in adult off-
programming of offspring emotionality after spring through methyl supplementation: alter-
stress early in pregnancy. J. Neurosci. 28: 9055– ing epigenetic marking later in life. J. Neurosci.
9065. 25: 11045–11054.
3. Tsankova, N.M. et al. 2006. Sustained hip- 15. Grayson, D.R. et al. 2005. Reelin promoter hy-
pocampal chromatin regulation in a mouse permethylation in schizophrenia. Proc. Natl.
model of depression and antidepressant ac- Acad. Sci. USA 102: 9341–9346.
tion. Nat. Neurosci. 9: 519–525. 16. McGowan, P.O. et al. 2008. Promoter-wide hy-
4. Lubin, F.D., T.L. Roth & J.D. Sweatt. 2008. Epi- permethylation of the ribosomal RNA gene
genetic regulation of bdnf gene transcription promoter in the suicide brain. PLoS ONE 3:
in the consolidation of fear memory. J. Neu- e2085.
rosci. 28: 10576–10586. 17. Miller, C.A. & J.D. Sweatt. 2007. Covalent
5. Jakobsson, J. et al. 2008. KAP1-mediated epi- modification of DNA regulates memory for-
genetic repression in the forebrain modulates mation. Neuron 53: 857–869.
behavioral vulnerability to stress. Neuron 60: 18. Korzus, E., M.G. Rosenfeld, M. Mayford. 2004.
818–831. CBP histone acetyltransferase activity is a criti-
6. McGowan, P.O. et al. 2009. Epigenetic regu- cal component of memory consolidation. Neu-
lation of the glucocorticoid receptor in hu- ron 42: 961–972.

Ann. N.Y. Acad. Sci. 1226 (2011) 14–33 


c 2011 New York Academy of Sciences. 29
Behavioral epigenetics Lester et al.

19. Levenson, J.M. et al. 2004. Regulation of his- implications for the involvement of epigenetic
tone acetylation during memory formation in processes. Biol. Psychiatr. 64: 645–652.
the hippocampus. J. Biol. Chem. 279: 40545– 30. Bönsch, D. et al. 2006. Lowered DNA methyl-
40559. transferase (DNMT-3b) mRNA expression is
20. Weaver, I.C.G. et al. 2007. The transcription associated with genomic DNA hypermethy-
factor nerve growth factor-inducible protein lation in patients with chronic alcoholism.
A mediates epigenetic programming: altering J. Neural Trans. 113: 1299–1304.
epigenetic marks by immediate-early genes. 31. Onishchenko, N., N. Karpova, F. Sabri, et al.
J. Neurosci. 27: 1756–1768. 2008. Long-lasting depression-like behavior
21. Chandramohan, Y., S.K. Droste, J.S. Arthur, and epigenetic changes of BDNF gene expres-
& J.M. Reul. 2008. The forced swimming- sion induced by perinatal exposure to methyl-
induced behavioural immobility response mercury. J. Neurochem. 106: 1378–1387.
involves histone H3 phospho-acetylation and 32. Frieling, H. et al. 2008. Epigenetic downreg-
c-Fos induction in dentate gyrus granule ulation of atrial natriuretic peptide but not
neurons via activation of the N -methyl- vasopressin mRNA expression in females with
D-aspartate/extracellular signal-regulated eating disorders is related to impulsivity. Neu-
kinase/mitogen- and stress-activated kinase ropsychopharmacology 33: 2605–2609.
signalling pathway. Eur. J. Neurosci. 27: 33. Philibert, R.A. et al. 2008. The relationship of
2701–2713. 5HTT (SLC6A4) methylation and genotype on
22. Collins, A. et al. 2009. Exercise improves cogni- mRNA expression and liability to major de-
tive responses to psychological stress through pression and alcohol dependence in subjects
enhancement of epigenetic mechanisms and from the Iowa Adoption Studies. Am. J. Med.
gene expression in the dentate gyrus. PLoS Genet. B. Neuropsychiatr. Genet. 147B: 543–
ONE 4: e4330. 549.
23. Roth, T.L. et al. 2009. Lasting epigenetic influ- 34. Nagarajan, R.P. et al. 2008. MECP2 Promoter
ence of early-life adversity on the BDNF gene. methylation and X chromosome inactivation
Biol. Psychiatr. 65: 760–769. in autism. Autism Res. 1: 169–178.
24. Ernst, C. et al. 2009. Alternative splicing, 35. Levine, A.A. et al. 2005. CREB-binding pro-
methylation state, and expression profile of tein controls response to cocaine by acetylat-
tropomyosin-related kinase B in the frontal ing histones at the fosB promoter in the mouse
cortex of suicide completers. Arch. Gen. Psy- striatum. PNAS 102: 19186–19191.
chiatr. 66: 22–32. 36. Breton, C.V. et al. 2009. Prenatal tobacco
25. Alter, M.D. et al. 2008. Variation in the large- smoke exposure affects global and gene-
scale organization of gene expression levels in specific DNA methylation. Am. J. Respir. Crit.
the hippocampus relates to stable epigenetic Care Med. 180: 462–467.
variability in behavior. PLoS ONE 3: e3344. 37. Mill, J. et al. 2008. Epigenomic profiling
26. Romieu, P. 2008. Histone deacetylase in- reveals DNA-methylation changes associated
hibitors decrease cocaine but not sucrose self- with major psychosis. Am. J. Hum. Genet. 82:
administration in rats. J. Neurosci. 28: 9342– 696–711.
9348. 38. Philibert, R.A., T.D. Gunter, S.R.H. Beach, et al.
27. Cassel, S. et al. 2006. Fluoxetine and cocaine in- 2008. MAOA methylation is associated with
duce the epigenetic factors MeCP2 and MBD1 nicotine and alcohol dependence in women.
in adult rat brain. Mol. Pharmacol. 70: 487– Am. J. Med. Genet. Pt. B. Neuropsychiatr. Genet.
492. Vol. 147B 5: 565–570.
28. Renthal, W. et al. 2007. Histone deacetylase 5 39. Wang, S.C., B. Oelze & A. Schumacher.
epigenetically controls behavioral adaptations 2008. Age-specific epigenetic drift in late-
to chronic emotional stimuli. Neuron 56: 517– onset alzheimer’s disease. PLoS One 16: e2698.
529. 40. Chwang, W.B., J.S. Arthur, A. Schumacher, &
29. Poulter, M.O. et al. 2008. GABAA receptor J.D. Sweatt. 2007. The nuclear kinase mitogen-
promoter hypermethylation in suicide brain: and stress-activated protein kinase 1 regulates

30 Ann. N.Y. Acad. Sci. 1226 (2011) 14–33 


c 2011 New York Academy of Sciences.
Lester et al. Behavioral epigenetics

hippocampal chromatin remodeling in mem- 50. Klempan, T.A., Ernst, C., Deleva, et al. 2009.
ory formation. J. Neurosci.27: 12732–12742. Characterization of QKI gene expression, ge-
41. Iwamoto, K. et al. 2005. DNA methylation netics, and epigenetics in suicide victims with
status of SOX10 correlates with its downreg- major depressive disorder. Biol. Psychiatry 66:
ulation and oligodendrocyte dysfunction in 824–831.
schizophrenia. J. Neurosci. 1: 5376–5381. 51. Huang, H.S. et al. 2007. Prefrontal dysfunc-
42. Champagne, F.A. et al. 2006. Maternal care tion in schizophrenia involves mixed-lineage
associated with methylation of the estro- leukemia 1-regulated histone methylation at
gen receptor-alpha1b promoter and estrogen GABAergic gene promoters. J. Neurosci. 27:
receptor-alpha expression in the medial pre- 11254–11262.
optic area of female offspring. Endocrinology 52. Huang, H.S. & S. Akbarian. 2007. GAD1
147: 2909–2915. mRNA expression and DNA methylation in
43. Nielsen, D.A. et al. 2009. Increased OPRM1 prefrontal cortex of subjects with schizophre-
DNA methylation in lymphocytes of nia. PLoS One 2: e809
methadone-maintained former heroin ad- 53. Deutsch, S.I. et al. 2008. Sodium butyrate, an
dicts. Neuropsychopharmacology 34: 867–873. epigenetic interventional strategy, attenuates
44. Bönsch, D., B. Lenz, J. Kornhuber & S. a stress-induced alteration of MK-801’s phar-
Bleich. 2005. DNA hypermethylation of the macologic action. Eur. Neuropsychopharmacol.
alpha synuclein promoter in patients with al- 8: 565–568.
coholism. Neuroreport. 8: 167–170. 54. Ferrante, R.J. et al. 2003. Histone deacetylase
45. Bleich, S. et al. 2006. Epigenetic DNA hy- inhibition by sodium butyrate chemotherapy
permethylation of the HERP gene promoter ameliorates the neurodegenerative phenotype
induces down-regulation of its mRNA expres- in Huntington’s disease mice. J. Neurosci. 23:
sion in patients with alcohol dependence. Al- 9418–9427.
cohol Clin. Exp. Res. 30: 587–591. 55. Bredy, T.W. et al. 2007. Histone modifications
46. Bönsch, D., B. Lenz, U. Reulbach, et al. around individual BDNF gene promoters in
2004. Homocysteine associated genomic DNA prefrontal cortex are associated with extinc-
hypermethylation in patients with chronic tion of conditioned fear. Learn. Mem. 14: 268–
alcoholism. J. Neura.l Transm. 111: 1611– 276.
1616. 56. Guipponi, M. et al. 2009. Genetic and epige-
47. Sathyan, P., H.B. Golden & R.C. Miranda. netic analysis of SSAT gene dysregulation in
2007. Competing interactions between micro- suicidal behavior. Am. J. Med. Genet. Pt B: Neu-
RNAs determine neural progenitor survival ropsychiatr. Genet. Vol. 150B 6: 799–807.
and proliferation after ethanol exposure: ev- 57. Pascual, M., J. Boix, V. Felipo & C. Guerri.
idence from an ex vivo model of the fetal cere- 2009. Repeated alcohol administration during
bral cortical neuroepithelium. J. Neurosci. 8: adolescence causes changes in the mesolim-
8546–8557. bic dopaminergic and glutamatergic systems
48. Daniels, W.M. et al. 2009. Maternal separa- and promotes alcohol intake in the adult rat.
tion alters nerve growth factor and corticos- J. Neurochem. 108: 920–931.
terone levels but not the DNA methylation 58. Renthal, W. et al. 2008. Delta FosB mediates
status of the exon 1(7) glucocorticoid receptor epigenetic desensitization of the c-fos gene af-
promoter region. Metab. Brain Dis. 24: 615– ter chronic amphetamine exposure. J. Neu-
627. rosci. 28: 7344–7349.
49. Fuchikamia, M., S. Morinobua, A. Kurataa1, 59. Hillemacher, T. et al. 2009. Promoter specific
S. Yamamotoa & S. Yamawakia. 2009. Single methylation of the dopamine transporter gene
immobilization stress differentially alters the is altered in alcohol dependence and associated
expression profile of transcripts of the brain- with craving. J. Psychiatr. Res. 43: 388–392.
derived neurotrophic factor (BDNF) gene and 60. Burmistrova, O.A. et al. 2007. MicroRNA in
histone acetylation at its promoters in the rat schizophrenia: genetic and expression analysis
hippocampus. Int. J. Neuropsychopharmacol. of miR-130b (22q11). Biochemistry (Mosc) 72:
12: 73–82. 578–582.

Ann. N.Y. Acad. Sci. 1226 (2011) 14–33 


c 2011 New York Academy of Sciences. 31
Behavioral epigenetics Lester et al.

61. Azami, S. et al. 2006. Altered gene activity cor- 72. Plagge, A. et al. 2004. The imprinted signaling
related with long-term memory formation of protein XL alpha s is required for postnatal
conditioned taste aversion in Lymnaea. J. Neu- adaptation to feeding. Nat. Genet. 36: 818–826.
rosci. Res. 15: 1610–1620. 73. Fernández-Gonzalez, R. et al. 2004. Long-term
62. Marutha Ravindran, C.R. & M.K. Ticku. 2009. effect of in vitro culture of mouse embryos
Effect of 5-azacytidine on the methylation as- with serum on mRNA expression of imprint-
pects of NMDA receptor NR2B gene in the ing genes, development, and behavior. Proc.
cultured cortical neurons of mice. Neurochem. Natl. Acad. Sci. USA 101: 5880–5885.
Res. 34: 342–350. 74. Miura, K. et al. 2002. Neurobehavioral
63. Baek, M.N. et al. 2010. Artificial microRNA- and electroencephalographic abnormalities in
based neurokinin-1 receptor gene silencing re- Ube3a maternal-deficient mice. Neurobiol. Dis.
duces alcohol consumption in mice. Neurosci. 9: 149–159.
Lett. 475: 124–128. 75. Lefebvre, L. et al. 1998. Abnormal maternal
64. Wang, J. et al. 2010. CBP histone acetyl- behaviour and growth retardation associated
transferase activity regulates embryonic neu- with loss of the imprinted gene Mest. Nat.
ral differentiation in the normal and Genet. 20: 163–169.
Rubinstein-Taybi syndrome brain. Develop- 76. Peleg, S. et al. 2010. Altered histone acetyla-
mental Cell 18: 114–125. tion is associated with age-dependent mem-
65. Kaminen-Ahola, N. et al. 2010. Maternal ory impairment in mice. Science 328: 753–
ethanol consumption alters the epigenotype 756.
and the phenotype offspring in a mouse 77. Tochigi, M. et al. 2008. Methylation status
model. PLoS Genet. 6: e1000811. of the reelin promoter region in the brain
66. Dash, P.K., S.A. Orsi & A.N. Moore. 2009. of schizophrenic patients. Biol. Psychiatr. 63:
Histone deactylase inhibition combined with 530–533.
behavioral therapy enhances learning and 78. Bromberg, A., Y. Bersudsky, J. Levine & G.
memory following traumatic brain injury. Agam. 2009. Global leukocyte DNA methyla-
Neuroscience 163: 1–8. tion is not altered in euthymic bipolar patients.
67. Hager, R., J.M. Cheverud & J.B. Wolf. 2009. J. Affect. Disord. 118: 234–239.
Change in maternal environment induced by 79. Davies, W. et al. 2005. Xlr3b is a new imprinted
cross-fostering alters genetic and epigenetic ef- candidate for X-linked parent-of-origin effects
fects on complex traits in mice. Proc. Biol. Sci. on cognitive function in mice. Nat. Genet. 37:
276: 2949–2954. 625–639.
68. Doe, C.M. et al. 2009. Loss of the imprinted 80. Zhao, Z., L. Fan & K.M. Frick. 2010. Epi-
snoRNA mbii-52 leads to increased 5htr2c pre- genetic alterations regulate estradiol-induced
RNA editing and altered 5HT2CR-mediated enhancement of memory consolidation. Proc.
behaviour. Hum. Mol. Genet. 18: 2140–2148. Natl. Acad. Sci. USA. 23: 5605–5610.
69. Esler, M. et al. 2008. Human sympathetic nerve 81. Lee, C.T., Y.L. Ma & E.H. Lee. 2007.
biology: parallel influences of stress and epi- Serum- and glucocorticoid-inducible kinase1
genetics in essential hypertension and panic enhances contextual fear memory formation
disorder. Ann. N.Y. Acad. Sci.1148: 338–348. through down-regulation of the expression of
70. Lasek, A.W., Janak, P.H., L. He. 2007. Down- Hes5. J. Neurochem. 100: 1531–1542.
regulation of mu opioid receptor by RNA in- 82. Olsson, C.A. et al. 2010. Prospects for epige-
terference in the ventral tegmental area reduces netic research within cohort studies of psy-
ethanol consumption in mice. Genes Brain Be- chological disorder: a pilot investigation of a
hav. 6: 728–735. peripheral cell marker of epigenetic risk for
71. Liljelund, P., A. Handforth, G.E. Homanics & depression. Biol. Psychol. 83: 159–165.
R.W. Olsen. 2005. GABAA receptor beta3 sub- 83. Hillemacher, T. et al. 2009. Epigenetic reg-
unit gene-deficient heterozygous mice show ulation and gene expression of vasopressin
parent-of-origin and gender-related differ- and atrial natriuretic peptide in alcohol with-
ences in beta3 subunit levels, EEG, and behav- drawal. Psychoneuroendocrinology 34: 555–
ior. Brain Res. Dev. Brain Res. 157: 150–161. 560.

32 Ann. N.Y. Acad. Sci. 1226 (2011) 14–33 


c 2011 New York Academy of Sciences.
Lester et al. Behavioral epigenetics

84. de Mooij-van Malsen, J.G., H.A. van Lith, H. activity of APP23 mice in a novel environment
Oppelaar et al. 2009. Evidence for epigenetic is reversed by siRNA. Brain Res. 1243: 124–
interactions for loci on mouse chromosome 1 133.
regulating open field activity. Behav. Genet. 39: 91. Sharma, R.P., D.R. Grayson, & D.P. Gavin.
176–182. 2008. Histone deactylase 1 expression is in-
85. Ouko, L.A. et al. 2009. Effect of alcohol con- creased in the prefrontal cortex of schizophre-
sumption on CpG methylation in the differen- nia subjects: analysis of the National Brain
tially methylated regions of H19 and IG-DMR Databank microarray collection. Schizophr.
in male gametes: implications for fetal alcohol Res. 98: 111–117.
spectrum disorders. Alcohol Clin. Exp. Res. 33: 92. DiFiglia, M. et al. 2007. Therapeutic silencing
1615–1627. of mutant huntington with siRNA attenuates
86. Phiel, C.J. et al. 2001. Histone deacetylase striatal and cortical neuropathology and be-
is a direct target of valproic acid, a potent havioral deficits. Proc. Natl. Acad. Sci. USA.
anticonvulsant, mood stabilizer, and terato- 104: 17204–17209.
gen. J. Biol. Chem. 28: 36734–36741. 93. Itaba-Matsumoto, N. et al. 2007. Imprinting
87. Skinner, M.K., M.D. Anway, M.I. Savenkova, status of paternally imprinted DLX5 gene in
et al.2008. Transgenerational epigenetic pro- Japanese patients with Rett syndrome. Brain
gramming of the brain transcriptome and anx- Develop. 29: 491–495.
iety behavior. PLoS One 3: e3745. 94. Uddin, M. et al. 2010. Epigenetic and im-
88. Chertkow-Deutsher, Y., H. Cohen, E. Klein & mune function profiles associated with post-
D. Ben-Shachar. 2010. DNA methylation in traumatic stress disorder. Proc. Natl. Acad. Sci.
vulnerability to post-traumatic stress in rats: USA 18: 9470–9475.
evidence for the role of the postsynaptic den- 95. Marutha Ravindran, C.R., & M.K. Ticku. 2005.
sity protein Dlgap2. Int. J. Neuropsychophar- Role of CpG islands in the up-regulation of
macol. 3: 347–359. NMDA receptor NR2B gene expression fol-
89. Dzitoyeva, S., N. Dimitrijevic & H. Manev. lowing chronic ethanol treatment of cultured
2003. Gamma-aminobutyric acid B receptor 1 cortical neurons of mice. Neurochem. Int. 46:
mediates behavior-impairing actions of alco- 313–327.
hol in Drosophila: adult RNA interference and 96. Hunter, R.G., K.J. McCarthy, T.A. Milne, et al.
pharmacological evidence. Proc. Natl. Acad. 2009. Regulation of hippocampal H3 histone
Sci. USA 29: 5485–5490. methylation by acute and chronic stress. Proc.
90. Senechal, Y. et al. 2008. Increased exploratory Natl. Acad. Sci. USA 106: 20912–20917.

Ann. N.Y. Acad. Sci. 1226 (2011) 14–33 


c 2011 New York Academy of Sciences. 33

You might also like